151
|
Quan W, Kim HK, Moon EY, Kim SS, Choi CS, Komatsu M, Jeong YT, Lee MK, Kim KW, Kim MS, Lee MS. Role of hypothalamic proopiomelanocortin neuron autophagy in the control of appetite and leptin response. Endocrinology 2012; 153:1817-26. [PMID: 22334718 DOI: 10.1210/en.2011-1882] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Autophagy is a catabolic cellular process involving the degradation of the cell's own components. Although the role of autophagy of diverse tissues in body metabolism has been investigated, the importance of autophagy in hypothalamic proopiomelanocortin (POMC) neurons, key regulators of energy balance, has not been addressed. The role of autophagy in leptin sensitivity that is critical for the control of body weight and appetite has also not been investigated. We produced mice with specific deletion of autophagy-related 7 (Atg7), an essential autophagy gene, in hypothalamic POMC neurons (Atg7(ΔPOMC) mice). Atg7 expression was deficient in the arcuate nucleus of the hypothalamus of Atg7(ΔPOMC) mice. p62, a specific substrate of autophagy, accumulated in the hypothalamus of Atg7(ΔPOMC) mice, which colocalized with ubiquitin. Atg7(ΔPOMC) mice had increased body weight due to increased food intake and decreased energy expenditure. Atg7(ΔPOMC) mice were not more prone to diet-induced obesity compared with control mice but more susceptible to hyperglycemia after high-fat diet. The ability of leptin to suppress fasting-elicited hyperphagia and weight gain during refeeding was attenuated in Atg7(ΔPOMC) mice. Deficient autophagy did not significantly affect POMC neuron number but impaired leptin-induced signal transducer and activation of transcription 3 activation. Our findings indicate a critical role for autophagy of POMC neurons in the control of energy homeostasis and leptin signaling.
Collapse
Affiliation(s)
- Wenying Quan
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
152
|
Villalba JM, de Cabo R, Alcain FJ. A patent review of sirtuin activators: an update. Expert Opin Ther Pat 2012; 22:355-67. [PMID: 22475539 DOI: 10.1517/13543776.2012.669374] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Reversible acetylation is a key post-translational modification of target proteins. Sirtuin deacetylases represent the homolog of the yeast silent information regulator (SIR2). Although seven sirtuins have been found in mammals, all sirtuin activators described to date act through SIRT1. AREAS COVERED Areas covered in this paper include a review of the patent literature associated with SIRT1 activators, with a focus on therapeutic applications, primarily related to the use of pharmaceuticals and nutraceuticals containing resveratrol (RSV), and the development of second-generation activators unrelated to RSV. Also discussed is the current controversy over whether or not these molecules are actual SIRT1 activators. EXPERT OPINION Developing effective strategies to protect against diet-induced metabolic imbalance is necessary to fight against current obesity rates. The hypothalamus is a candidate for developing drugs that suppress SIRT1 degradation, as a strategy for treating metabolic syndrome. Deciphering the basic mechanism of activators is essential to develop effective strategies to alter sirtuin activity. This is true regardless of the apparent controversy of whether in vitro activation of SIRT1 is direct or not, depending on the experimental design, and whether sirtuins may play a major role in longevity. The numerous studies on their positive effects against age-related diseases, obesity and other metabolic disorders are still valid, promising to positively influence the development of treatments to improve human health.
Collapse
Affiliation(s)
- José M Villalba
- Departamento de Biología Celular, Fisiología e Inmunología, Facultad de Ciencias, Campus Universitario de Rabanales, Universidad de Córdoba, Spain
| | | | | |
Collapse
|
153
|
Kaushik S, Arias E, Kwon H, Lopez NM, Athonvarangkul D, Sahu S, Schwartz GJ, Pessin JE, Singh R. Loss of autophagy in hypothalamic POMC neurons impairs lipolysis. EMBO Rep 2012; 13:258-65. [PMID: 22249165 DOI: 10.1038/embor.2011.260] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 12/16/2011] [Accepted: 12/16/2011] [Indexed: 11/09/2022] Open
Abstract
Autophagy degrades cytoplasmic contents to achieve cellular homeostasis. We show that selective loss of autophagy in hypothalamic proopiomelanocortin (POMC) neurons decreases α-melanocyte-stimulating hormone (MSH) levels, promoting adiposity, impairing lipolysis and altering glucose homeostasis. Ageing reduces hypothalamic autophagy and α-MSH levels, and aged-mice phenocopy, the adiposity and lipolytic defect observed in POMC neuron autophagy-null mice. Intraperitoneal isoproterenol restores lipolysis in both models, demonstrating normal adipocyte catecholamine responsiveness. We propose that an unconventional, autophagosome-mediated form of secretion in POMC neurons controls energy balance by regulating α-MSH production. Modulating hypothalamic autophagy might have implications for preventing obesity and metabolic syndrome of ageing.
Collapse
Affiliation(s)
- Susmita Kaushik
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Patel PR, Ramakrishnan SK, Kaw MK, Raphael CK, Ghosh S, Marino JS, Heinrich G, Lee SJ, Bourey RE, Hill JW, Jung DY, Morgan DA, Kim JK, Rahmouni SK, Najjar SM. Increased metabolic rate and insulin sensitivity in male mice lacking the carcino-embryonic antigen-related cell adhesion molecule 2. Diabetologia 2012; 55:763-72. [PMID: 22159884 PMCID: PMC3272352 DOI: 10.1007/s00125-011-2388-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 11/07/2011] [Indexed: 10/14/2022]
Abstract
AIMS/HYPOTHESIS The carcino-embryonic antigen-related cell adhesion molecule (CEACAM)2 is produced in many feeding control centres in the brain, but not in peripheral insulin-targeted tissues. Global Ceacam2 null mutation causes insulin resistance and obesity resulting from hyperphagia and hypometabolism in female Ceacam2 homozygous null mutant mice (Cc2 [also known as Ceacam2](-/-)) mice. Because male mice are not obese, the current study examined their metabolic phenotype. METHODS The phenotype of male Cc2(-/-) mice was characterised by body fat composition, indirect calorimetry, hyperinsulinaemic-euglycaemic clamp analysis and direct recording of sympathetic nerve activity. RESULTS Despite hyperphagia, total fat mass was reduced, owing to the hypermetabolic state in male Cc2(-/-) mice. In contrast to females, male mice also exhibited insulin sensitivity with elevated β-oxidation in skeletal muscle, which is likely to offset the effects of increased food intake. Males and females had increased brown adipogenesis. However, only males had increased activation of sympathetic tone regulation of adipose tissue and increased spontaneous activity. The mechanisms underlying sexual dimorphism in energy balance with the loss of Ceacam2 remain unknown. CONCLUSIONS/INTERPRETATION These studies identified a novel role for CEACAM2 in the regulation of metabolic rate and insulin sensitivity via effects on brown adipogenesis, sympathetic nervous outflow to brown adipose tissue, spontaneous activity and energy expenditure in skeletal muscle.
Collapse
Affiliation(s)
- P. R. Patel
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - S. K. Ramakrishnan
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - M. K. Kaw
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - C. K. Raphael
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - S. Ghosh
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - J. S. Marino
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - G. Heinrich
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - S. J. Lee
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - R. E. Bourey
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Internal Medicine at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - J. W. Hill
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - D. Y. Jung
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - D. A. Morgan
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - J. K. Kim
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - S. K. Rahmouni
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - S. M. Najjar
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA,
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| |
Collapse
|
155
|
Neri C. Role and Therapeutic Potential of the Pro-Longevity Factor FOXO and Its Regulators in Neurodegenerative Disease. Front Pharmacol 2012; 3:15. [PMID: 22363285 PMCID: PMC3281233 DOI: 10.3389/fphar.2012.00015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 01/25/2012] [Indexed: 12/25/2022] Open
Abstract
Studies in simple model organisms have yielded crucial insights into the genetic and molecular aspects of longevity. FOXO, which is most notable for its association with longevity, and its upstream regulators such as sirtuins have received particular attention in translational research because these genes modulate cell survival in several models of neurodegenerative diseases. There is a large amount of knowledge on the pathways that regulate FOXO activity and genes that may be regulated by FOXO. However, for the same reason that the FOXO network is a complex stress response system, its therapeutic potential to develop disease-modifying strategies requires further examination. Although the FOXO network contains druggable genes such as sirtuins and AMPK, whether they should be activated or inhibited and whether protection against the early or late phases of neuronal cell decline might require opposite therapeutic strategies remains unclear. Additionally, the mode of action of small compound molecules believed to act on FOXO network targets was questioned. This review recapitulates essential facts and questions about the promises of FOXO and its interactors in neurodegenerative disease.
Collapse
Affiliation(s)
- Christian Neri
- Laboratory of Neuronal Cell Biology and Pathology, Unit 894, INSERM Paris, France
| |
Collapse
|
156
|
Loss of autophagy in pro-opiomelanocortin neurons perturbs axon growth and causes metabolic dysregulation. Cell Metab 2012; 15:247-55. [PMID: 22285542 PMCID: PMC3278575 DOI: 10.1016/j.cmet.2011.12.016] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 10/24/2011] [Accepted: 12/06/2011] [Indexed: 12/17/2022]
Abstract
The hypothalamic melanocortin system, which includes neurons that produce pro-opiomelanocortin (POMC)-derived peptides, is a major negative regulator of energy balance. POMC neurons begin to acquire their unique properties during neonatal life. The formation of functional neural systems requires massive cytoplasmic remodeling that may involve autophagy, an important intracellular mechanism for the degradation of damaged proteins and organelles. Here we investigated the functional and structural effects of the deletion of an essential autophagy gene, Atg7, in POMC neurons. Lack of Atg7 in POMC neurons caused higher postweaning body weight, increased adiposity, and glucose intolerance. These metabolic impairments were associated with an age-dependent accumulation of ubiquitin/p62-positive aggregates in the hypothalamus and a disruption in the maturation of POMC-containing axonal projections. Together, these data provide direct genetic evidence that Atg7 in POMC neurons is required for normal metabolic regulation and neural development, and they implicate hypothalamic autophagy deficiency in the pathogenesis of obesity.
Collapse
|
157
|
Abstract
Metabolic diseases are an increasing threat in developed countries. Dysregulation of metabolic pathways, caused by imbalances in energy homeostasis, leads to obesity, diabetes and cardiovascular disease with devastating results for both individuals and societies. Sirtuins, a conserved family of NAD(+)-dependent deacetylase enzymes found in many species, regulate various metabolic pathways and have emerged as important sensors of energy status in mammals. The nuclear sirtuins, SIRT1, SIRT6 and SIRT7, regulate the activity of key transcription factors and cofactors of numerous metabolic pathways in almost all tissues by linking nutrient signals with the cellular responses to energy demands. The mitochondrial sirtuins, SIRT3, SIRT4 and SIRT5, regulate the activity of important mitochondrial enzymes and drive metabolic cycles in response to fasting and calorie restriction. Accumulating evidence indicates that sirtuins can be beneficial in the prevention of metabolic and age-related diseases and suggests that they can be pharmacologically activated to ameliorate such diseases. This Review describes the latest advances in the understanding of the function of sirtuins as regulators of mammalian metabolism and focuses on the role of these enzymes as mediators of nutrient availability.
Collapse
Affiliation(s)
- Angeliki Chalkiadaki
- Paul F. Glenn Laboratory, Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Building 68, Cambridge, MA 02139, USA
| | | |
Collapse
|
158
|
Chong ZZ, Shang YC, Wang S, Maiese K. SIRT1: new avenues of discovery for disorders of oxidative stress. Expert Opin Ther Targets 2012; 16:167-78. [PMID: 22233091 DOI: 10.1517/14728222.2012.648926] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The sirtuin SIRT1 is expressed throughout the body, has broad biological effects and can significantly affect both cellular survival and longevity during acute and long-term injuries, which involve both oxidative stress and cell metabolism. AREAS COVERED SIRT1 has an intricate role in the pathology, progression, and treatment of several disease entities, including neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease, tumorigenesis, cardiovascular disease with myocardial injury and atherosclerosis, metabolic disease, and aging-related disease. New areas of study in these disciplines, with discussion of the cellular biology, are highlighted. EXPERT OPINION Novel signaling pathways for SIRT1, which can be targeted to enhance cellular protection and potentially extend lifespan, continue to emerge. Investigations that can further determine the intracellular signaling, trafficking and post-translational modifications that occur with SIRT1 in a variety of cell systems and environments will allow us to further translate this knowledge into effective therapeutic strategies that will be applicable to multiple systems of the body.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- University of Medicine and Dentistry - New Jersey Medical School, Laboratory of Cellular and Molecular Signaling, Newark, NJ 07101, USA
| | | | | | | |
Collapse
|
159
|
Haws ME, Kaeser PS, Jarvis DL, Südhof TC, Powell CM. Region-specific deletions of RIM1 reproduce a subset of global RIM1α(-/-) phenotypes. GENES BRAIN AND BEHAVIOR 2012; 11:201-13. [PMID: 22103334 PMCID: PMC3268893 DOI: 10.1111/j.1601-183x.2011.00755.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The presynaptic protein RIM1α mediates multiple forms of presynaptic plasticity at both excitatory and inhibitory synapses. Previous studies of mice lacking RIM1α (RIM1α(-/-) throughout the brain showed that deletion of RIM1α results in multiple behavioral abnormalities. In an effort to begin to delineate the brain regions in which RIM1 deletion mediates these abnormal behaviors, we used conditional (floxed) RIM1 knockout mice (fRIM1). By crossing these fRIM1 mice to previously characterized transgenic cre lines, we aimed to delete RIM1 selectively in the dentate gyrus (DG), using a specific preproopiomelanocortin promoter driving cre recombinase (POMC-cre) line , and in pyramidal neurons of the CA3 region of hippocampus, using the kainate receptor subunit 1 promoter driving cre recombinase (KA-cre). Neither of these cre driver lines was uniquely selective to the targeted regions. In spite of this, we were able to reproduce a subset of the global RIM1α(-/-) behavioral abnormalities, thereby narrowing the brain regions in which loss of RIM1 is sufficient to produce these behavioral differences. Most interestingly, hypersensitivity to the pyschotomimetic MK-801 was shown in mice lacking RIM1 selectively in the DG, arcuate nucleus of the hypothalamus and select cerebellar neurons, implicating novel brain regions and neuronal subtypes in this behavior.
Collapse
Affiliation(s)
- M E Haws
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | |
Collapse
|
160
|
Townsend K, Tseng YH. Brown adipose tissue: Recent insights into development, metabolic function and therapeutic potential. Adipocyte 2012; 1:13-24. [PMID: 23700507 PMCID: PMC3661118 DOI: 10.4161/adip.18951] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Obesity is currently a global pandemic, and is associated with increased mortality and co-morbidities including many metabolic diseases. Obesity is characterized by an increase in adipose mass due to increased energy intake, decreased energy expenditure, or both. While white adipose tissue is specialized for energy storage, brown adipose tissue has a high concentration of mitochondria and uniquely expresses uncoupling protein 1, enabling it to be specialized for energy expenditure and thermogenesis. Although brown fat was once considered only necessary in babies, recent morphological and imaging studies have provided evidence that, contrary to prior belief, this tissue is present and active in adult humans. In recent years, the topic of brown adipose tissue has been reinvigorated with many new studies regarding brown adipose tissue differentiation, function and therapeutic promise. This review summarizes the recent advances, discusses the emerging questions and offers perspective on the potential therapeutic applications targeting this tissue.
Collapse
Affiliation(s)
- Kristy Townsend
- Joslin Diabetes Center and Harvard Medical School; Boston, MA USA
| | - Yu-Hua Tseng
- Joslin Diabetes Center and Harvard Medical School; Boston, MA USA
- Harvard Stem Cell Institute; Harvard University; Cambridge, MA USA
| |
Collapse
|
161
|
Li K, Luo J, Luo J, Azabdaftari G. The role of SIRT1 in tumorigenesis. NORTH AMERICAN JOURNAL OF MEDICINE & SCIENCE 2011; 4:104-106. [PMID: 22180829 DOI: 10.7156/v4i2p104] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
NAD-dependent Class III histone deacetylase SIRT1 is a multiple functional protein and has been demonstrated critically involved in stress response, cellular metabolism and aging through deacetylating variety of substrates including p53, forkhead transcription factors, PGC-1α, NF-κB, Ku70 and histones. Increasing evidences indicate that SIRT1 plays a complex role in tumorigenesis with functions in both tumor promoting and tumor suppressing. This review provides an overview of current knowledge of SIRT1 and its controversies regarding the functions of SIRT1 in tumorigenesis.
Collapse
Affiliation(s)
- Kai Li
- Department of Medical & Research Technology, Department of Pathology, School of Medicine, University of Maryland, AHB 405A, 100 Penn Street, Baltimore, MD 21201
| | | | | | | | | | | |
Collapse
|
162
|
Cohen-Kfir E, Artsi H, Levin A, Abramowitz E, Bajayo A, Gurt I, Zhong L, D'Urso A, Toiber D, Mostoslavsky R, Dresner-Pollak R. Sirt1 is a regulator of bone mass and a repressor of Sost encoding for sclerostin, a bone formation inhibitor. Endocrinology 2011; 152:4514-24. [PMID: 21952235 DOI: 10.1210/en.2011-1128] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Sirt1, the mammalian ortholog of the yeast Sir2 (silent information regulator 2), was shown to play an important role in metabolism and in age-associated diseases, but its role in skeletal homeostasis and osteoporosis has yet not been studied. Using 129/Sv mice with a germline mutation in the Sirt1 gene, we demonstrate that Sirt1 haplo-insufficient (Sirt1(+/-)) female mice exhibit a significant reduction in bone mass characterized by decreased bone formation and increased marrow adipogenesis. Importantly, we identify Sost, encoding for sclerostin, a critical inhibitor of bone formation, as a novel target of Sirt1. Using chromatin immunoprecipitation analysis, we reveal that Sirt1 directly and negatively regulates Sost gene expression by deacetylating histone 3 at lysine 9 at the Sost promoter. Sost down-regulation by small interfering RNA and the administration of a sclerostin-neutralizing antibody restore gene expression of osteocalcin and bone sialoprotein as well as mineralized nodule formation in Sirt1(+/-) marrow-derived mesenchymal stem cells induced to osteogenesis. These findings reveal a novel role for Sirt1 in bone as a regulator of bone mass and a repressor of sclerostin, and have potential implications suggesting that Sirt1 is a target for promoting bone formation as an anabolic approach for treatment of osteoporosis.
Collapse
Affiliation(s)
- Einav Cohen-Kfir
- Department of Medicine, Endocrinology and Metabolism Service, Hadassah-Hebrew University Medical Center, P.O. Box 12000, Jerusalem 91120, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Lakhan SE, Kirchgessner A. Gut microbiota and sirtuins in obesity-related inflammation and bowel dysfunction. J Transl Med 2011; 9:202. [PMID: 22115311 PMCID: PMC3235071 DOI: 10.1186/1479-5876-9-202] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Accepted: 11/24/2011] [Indexed: 02/07/2023] Open
Abstract
Obesity is a chronic disease characterized by persistent low-grade inflammation with alterations in gut motility. Motor abnormalities suggest that obesity has effects on the enteric nervous system (ENS), which controls virtually all gut functions. Recent studies have revealed that the gut microbiota can affect obesity and increase inflammatory tone by modulating mucosal barrier function. Furthermore, the observation that inflammatory conditions influence the excitability of enteric neurons may add to the gut dysfunction in obesity. In this article, we discuss recent advances in understanding the role of gut microbiota and inflammation in the pathogenesis of obesity and obesity-related gastrointestinal dysfunction. The potential contribution of sirtuins in protecting or regulating the circuitry of the ENS under inflamed states is also considered.
Collapse
Affiliation(s)
- Shaheen E Lakhan
- Global Neuroscience Initiative Foundation, Los Angeles, CA, USA.
| | | |
Collapse
|
164
|
Knight CM, Gutierrez-Juarez R, Lam TKT, Arrieta-Cruz I, Huang L, Schwartz G, Barzilai N, Rossetti L. Mediobasal hypothalamic SIRT1 is essential for resveratrol's effects on insulin action in rats. Diabetes 2011; 60:2691-700. [PMID: 21896928 PMCID: PMC3198094 DOI: 10.2337/db10-0987] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Sirtuin 1 (SIRT1) and its activator resveratrol are emerging as major regulators of metabolic processes. We investigate the site of resveratrol action on glucose metabolism and the contribution of SIRT1 to these effects. Because the arcuate nucleus in the mediobasal hypothalamus (MBH) plays a pivotal role in integrating peripheral metabolic responses to nutrients and hormones, we examined whether the actions of resveratrol are mediated at the MBH. RESEARCH DESIGN AND METHODS Sprague Dawley (SD) male rats received acute central (MBH) or systemic injections of vehicle, resveratrol, or SIRT1 inhibitor during basal pancreatic insulin clamp studies. To delineate the pathway(s) by which MBH resveratrol modulates hepatic glucose production, we silenced hypothalamic SIRT1 expression using a short hairpin RNA (shRNA) inhibited the hypothalamic ATP-sensitive potassium (K(ATP)) channel with glibenclamide, or selectively transected the hepatic branch of the vagus nerve while infusing resveratrol centrally. RESULTS Our studies show that marked improvement in insulin sensitivity can be elicited by acute administration of resveratrol to the MBH or during acute systemic administration. Selective inhibition of hypothalamic SIRT1 using a cell-permeable SIRT1 inhibitor or SIRT1-shRNA negated the effect of central and peripheral resveratrol on glucose production. Blockade of the K(ATP) channel and hepatic vagotomy significantly attenuated the effect of central resveratrol on hepatic glucose production. In addition, we found no evidence for hypothalamic AMPK activation after MBH resveratrol administration. CONCLUSIONS Taken together, these studies demonstrate that resveratrol improves glucose homeostasis mainly through a central SIRT1-dependent pathway and that the MBH is a major site of resveratrol action.
Collapse
Affiliation(s)
- Colette M Knight
- Department of Medicine, Diabetes Research Center, Albert EinsteinCollege of Medicine, Bronx, New York, USA.
| | | | | | | | | | | | | | | |
Collapse
|
165
|
Abstract
The hedonic properties of food can stimulate feeding behaviour even when energy requirements have been met, contributing to weight gain and obesity. Similarly, the hedonic effects of drugs of abuse can motivate their excessive intake, culminating in addiction. Common brain substrates regulate the hedonic properties of palatable food and addictive drugs, and recent reports suggest that excessive consumption of food or drugs of abuse induces similar neuroadaptive responses in brain reward circuitries. Here, we review evidence suggesting that obesity and drug addiction may share common molecular, cellular and systems-level mechanisms.
Collapse
|
166
|
Purushotham A, Xu Q, Li X. Systemic SIRT1 insufficiency results in disruption of energy homeostasis and steroid hormone metabolism upon high-fat-diet feeding. FASEB J 2011; 26:656-67. [PMID: 22006157 DOI: 10.1096/fj.11-195172] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIRT1 is a highly-conserved NAD(+)-dependent protein deacetylase that plays essential roles in the regulation of energy metabolism, genomic stability, and stress response. Although the functions of SIRT1 in many organs have been extensively studied in tissue-specific knockout mouse models, the systemic role of SIRT1 is still largely unknown as a result of severe developmental defects that result from whole-body knockout in mice. Here, we investigated the systemic functions of SIRT1 in metabolic homeostasis by utilizing a whole-body SIRT1 heterozygous mouse model. These mice are phenotypically normal under standard feeding conditions. However, when chronically challenged with a 40% fat diet, they become obese and insulin resistant, display increased serum cytokine levels, and develop hepatomegaly. Hepatic metabolomic analyses revealed that SIRT1 heterozygous mice have elevated gluconeogenesis and oxidative stress. Surprisingly, they are depleted of glycerolipid metabolites and free fatty acids, yet accumulate lysolipids. Moreover, high-fat feeding induces elevation of serum testosterone levels and enlargement of seminal vesicles in SIRT1 heterozygous males. Microarray analysis of liver mRNA indicates that they have altered expression of genes involved in steroid metabolism and glycerolipid metabolism. Taken together, our findings indicate that SIRT1 plays a vital role in the regulation of systemic energy and steroid hormone homeostasis.
Collapse
Affiliation(s)
- Aparna Purushotham
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | | | | |
Collapse
|
167
|
Xu Y, Nedungadi TP, Zhu L, Sobhani N, Irani BG, Davis KE, Zhang X, Zou F, Gent LM, Hahner LD, Khan SA, Elias CF, Elmquist JK, Clegg DJ. Distinct hypothalamic neurons mediate estrogenic effects on energy homeostasis and reproduction. Cell Metab 2011; 14:453-65. [PMID: 21982706 PMCID: PMC3235745 DOI: 10.1016/j.cmet.2011.08.009] [Citation(s) in RCA: 450] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 06/24/2011] [Accepted: 08/02/2011] [Indexed: 11/24/2022]
Abstract
Estrogens regulate body weight and reproduction primarily through actions on estrogen receptor-α (ERα). However, ERα-expressing cells mediating these effects are not identified. We demonstrate that brain-specific deletion of ERα in female mice causes abdominal obesity stemming from both hyperphagia and hypometabolism. Hypometabolism and abdominal obesity, but not hyperphagia, are recapitulated in female mice lacking ERα in hypothalamic steroidogenic factor-1 (SF1) neurons. In contrast, deletion of ERα in hypothalamic pro-opiomelanocortin (POMC) neurons leads to hyperphagia, without directly influencing energy expenditure or fat distribution. Further, simultaneous deletion of ERα from both SF1 and POMC neurons causes hypometabolism, hyperphagia, and increased visceral adiposity. Additionally, female mice lacking ERα in SF1 neurons develop anovulation and infertility, while POMC-specific deletion of ERα inhibits negative feedback regulation of estrogens and impairs fertility in females. These results indicate that estrogens act on distinct hypothalamic ERα neurons to regulate different aspects of energy homeostasis and reproduction.
Collapse
Affiliation(s)
- Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Ramadori G, Fujikawa T, Anderson J, Berglund ED, Frazao R, Michán S, Vianna CR, Sinclair DA, Elias CF, Coppari R. SIRT1 deacetylase in SF1 neurons protects against metabolic imbalance. Cell Metab 2011; 14:301-12. [PMID: 21907137 PMCID: PMC3172583 DOI: 10.1016/j.cmet.2011.06.014] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 05/04/2011] [Accepted: 06/09/2011] [Indexed: 11/30/2022]
Abstract
Chronic feeding on high-calorie diets causes obesity and type 2 diabetes mellitus (T2DM), illnesses that affect hundreds of millions. Thus, understanding the pathways protecting against diet-induced metabolic imbalance is of paramount medical importance. Here, we show that mice lacking SIRT1 in steroidogenic factor 1 (SF1) neurons are hypersensitive to dietary obesity owing to maladaptive energy expenditure. Also, mutant mice have increased susceptibility to developing dietary T2DM due to insulin resistance in skeletal muscle. Mechanistically, these aberrations arise, in part, from impaired metabolic actions of the neuropeptide orexin-A and the hormone leptin. Conversely, mice overexpressing SIRT1 in SF1 neurons are more resistant to diet-induced obesity and insulin resistance due to increased energy expenditure and enhanced skeletal muscle insulin sensitivity. Our results unveil important protective roles of SIRT1 in SF1 neurons against dietary metabolic imbalance.
Collapse
Affiliation(s)
- Giorgio Ramadori
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
De Jonghe BC, Hayes MR, Bence KK. Melanocortin control of energy balance: evidence from rodent models. Cell Mol Life Sci 2011; 68:2569-88. [PMID: 21553232 PMCID: PMC3135719 DOI: 10.1007/s00018-011-0707-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 04/19/2011] [Accepted: 04/20/2011] [Indexed: 01/18/2023]
Abstract
Regulation of energy balance is extremely complex, and involves multiple systems of hormones, neurotransmitters, receptors, and intracellular signals. As data have accumulated over the last two decades, the CNS melanocortin system is now identified as a prominent integrative network of energy balance controls in the mammalian brain. Here, we will review findings from rat and mouse models, which have provided an important framework in which to study melanocortin function. Perhaps most importantly, this review attempts for the first time to summarize recent advances in our understanding of the intracellular signaling pathways thought to mediate the action of melanocortin neurons and peptides in control of longterm energy balance. Special attention will be paid to the roles of MC4R/MC3R, as well as downstream neurotransmitters within forebrain and hindbrain structures that illustrate the distributed control of melanocortin signaling in energy balance. In addition, distinctions and controversy between rodent species will be discussed.
Collapse
Affiliation(s)
- Bart C. De Jonghe
- Dept. of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Matthew R. Hayes
- Dept. of Psychiatry, School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Kendra K. Bence
- Dept. of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
170
|
Wu D, Qiu Y, Gao X, Yuan XB, Zhai Q. Overexpression of SIRT1 in mouse forebrain impairs lipid/glucose metabolism and motor function. PLoS One 2011; 6:e21759. [PMID: 21738790 PMCID: PMC3128079 DOI: 10.1371/journal.pone.0021759] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 06/06/2011] [Indexed: 11/18/2022] Open
Abstract
SIRT1 plays crucial roles in glucose and lipid metabolism, and has various functions in different tissues including brain. The brain-specific SIRT1 knockout mice display defects in somatotropic signaling, memory and synaptic plasticity. And the female mice without SIRT1 in POMC neuron are more sensitive to diet-induced obesity. Here we created transgenic mice overexpressing SIRT1 in striatum and hippocampus under the control of CaMKIIα promoter. These mice, especially females, exhibited increased fat accumulation accompanied by significant upregulation of adipogenic genes in white adipose tissue. Glucose tolerance of the mice was also impaired with decreased Glut4 mRNA levels in muscle. Moreover, the SIRT1 overexpressing mice showed decreased energy expenditure, and concomitantly mitochondria-related genes were decreased in muscle. In addition, these mice showed unusual spontaneous physical activity pattern, decreased activity in open field and rotarod performance. Further studies demonstrated that SIRT1 deacetylated IRS-2, and upregulated phosphorylation level of IRS-2 and ERK1/2 in striatum. Meanwhile, the neurotransmitter signaling in striatum and the expression of endocrine hormones in hypothalamus and serum T3, T4 levels were altered. Taken together, our findings demonstrate that SIRT1 in forebrain regulates lipid/glucose metabolism and motor function.
Collapse
Affiliation(s)
- Dongmei Wu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Yifu Qiu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Xiang Gao
- Model Animal Research Center of Nanjing University, Nanjing, China
| | - Xiao-Bing Yuan
- Institute of Neuroscience and State Key Laboratory of Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
171
|
Affiliation(s)
- Takashi Nakagawa
- Paul F. Glenn Laboratory for the Science of Aging, Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Building 68-280, Cambridge, Cambridge, MA 02139, USA
| | | |
Collapse
|
172
|
Affiliation(s)
- Leonard Guarente
- Paul F. Glenn Lab and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
173
|
Li X, Kazgan N. Mammalian sirtuins and energy metabolism. Int J Biol Sci 2011; 7:575-87. [PMID: 21614150 PMCID: PMC3101526 DOI: 10.7150/ijbs.7.575] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 03/30/2011] [Indexed: 12/11/2022] Open
Abstract
Sirtuins are highly conserved NAD+-dependent protein deacetylases and/or ADP-ribosyltransferases that can extend the lifespan of several lower model organisms including yeast, worms and flies. The seven mammalian sirtuins, SIRT1 to SIRT7, have emerged as key metabolic sensors that directly link environmental signals to mammalian metabolic homeostasis and stress response. Recent studies have shed light on the critical roles of sirtuins in mammalian energy metabolism in response to nutrient signals. This review focuses on the involvement of two nuclear sirtuins, SIRT1 and SIRT6, and three mitochondrial sirtuins, SIRT3, SIRT4, and SIRT5, in regulation of diverse metabolic processes.
Collapse
Affiliation(s)
- Xiaoling Li
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|
174
|
Imai SI. Dissecting systemic control of metabolism and aging in the NAD World: the importance of SIRT1 and NAMPT-mediated NAD biosynthesis. FEBS Lett 2011; 585:1657-62. [PMID: 21550345 DOI: 10.1016/j.febslet.2011.04.060] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 04/27/2011] [Accepted: 04/28/2011] [Indexed: 11/24/2022]
Abstract
Many countries are facing social and economic problems due to increased elderly demographics. With these demands, it is now critical to understand the fundamental regulatory mechanism for aging and longevity in mammals. Our studies on the mammalian NAD-dependent deacetylase SIRT1 and nicotinamide phosphoribosyltransferase (NAMPT)-mediated systemic NAD biosynthesis led us to propose a comprehensive model for the systemic regulatory network connecting metabolism and aging, termed the "NAD World". In this article, I will discuss the importance of SIRT1 and NAMPT-mediated NAD biosynthesis in the NAD World and the system dynamics of this hierarchical network for the connection between metabolism and aging.
Collapse
Affiliation(s)
- Shin-ichiro Imai
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
175
|
Tang BL. Sirt1's systemic protective roles and its promise as a target in antiaging medicine. Transl Res 2011; 157:276-84. [PMID: 21497775 DOI: 10.1016/j.trsl.2010.11.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 11/24/2010] [Accepted: 11/24/2010] [Indexed: 12/14/2022]
Abstract
Silent information regulator 2 (Sir2/Sirt1), a member of the sirtuin family of class III histone deacetylases, has been implicated extensively in lifespan extension and is a prominent drug target in antiaging medicine. The mammalian Sirt1 has multiple targets, which include histones, transcription factors, and other molecules that collectively modulate energy metabolism, stress response, and cell/tissue survival. Several of Sirt1's substrates regulate key metabolic processes, and Sirt1 activation may underlie the lifespan prolonging effect of caloric restriction. Recent studies have also identified multifaceted protective roles for Sirt1 against cellular senescence and stress in the neural, cardiovascular, and renal systems. Sirt1's activity in multiple tissues may decline with aging, and sustaining or reactivating this activity seems invariably beneficial. Several studies also point towards a general tumor suppressive role for Sirt1, at least in the context of certain human cancers. Development of Sirt1-based therapeutic interventions against systemic aging and aging-associated diseases will benefit from a thorough understanding of underlying pathological mechanisms of diseases as well as metabolic connections between different tissues and organs.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore 117597.
| |
Collapse
|
176
|
Abstract
Sirtuin 1 (SIRT1), the mammalian ortholog of yeast Sir2, is a highly conserved NAD(+)-dependent protein deacetylase that has emerged as a key metabolic sensor that directly links environmental nutrient signals to animal metabolic homeostasis. SIRT1 is known to be involved in gluconeogenesis in the liver, fat mobilization in white adipose tissue, and insulin secretion in the pancreas. Recent studies have shown SIRT1 to regulate fatty acid oxidation in the liver, sense nutrient availability in the hypothalamus, influence obesity-induced inflammation in macrophages, and modulate the activity of the circadian clock in metabolic tissues. The activity of SIRT1 also appears to be under the control of AMPK and adiponectin. This review focuses on the involvement of SIRT1 in regulating metabolic diseases associated with obesity. It includes brief overviews of sirtuin signaling, with emphasis on SIRT1's role in the liver, macrophage, brain, and adipose tissue as it relates to obesity.
Collapse
Affiliation(s)
- Thaddeus T Schug
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, RTP, NC 27709, USA
| | | |
Collapse
|
177
|
Begriche K, Sutton GM, Butler AA. Homeostastic and non-homeostatic functions of melanocortin-3 receptors in the control of energy balance and metabolism. Physiol Behav 2011; 104:546-54. [PMID: 21497617 DOI: 10.1016/j.physbeh.2011.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 04/04/2011] [Accepted: 04/08/2011] [Indexed: 01/05/2023]
Abstract
The central nervous melanocortin system is a neural network linking nutrient-sensing systems with hypothalamic, limbic and hindbrain neurons regulating behavior and metabolic homeostasis. Primary melanocortin neurons releasing melanocortin receptor ligands residing in the hypothalamic arcuate nucleus are regulated by nutrient-sensing and metabolic signals. A smaller group of primary neurons releasing melanocortin agonists in the nucleus tractus solitarius in the brainstem are also regulated by signals of metabolic state. Two melanocortin receptors regulate energy homeostasis. Melanocortin-4 receptors regulate satiety and autonomic outputs controlling peripheral metabolism. The functions of melanocortin-3 receptors (MC3R) expressed in hypothalamic and limbic structures are less clear. Here we discuss published data and preliminary observations from our laboratory suggesting that neural MC3R regulate inputs into systems governing the synchronization of rhythms in behavior and metabolism with nutrient intake. Mice subjected to a restricted feeding protocol, where a limited number of calories are presented at a 24h interval, rapidly exhibit bouts of increased wakefulness and activity which anticipate food presentation. The full expression of these responses is dependent on MC3R. Moreover, MC3R knockout mice are unique in exhibiting a dissociation of weight loss from improved glucose homeostasis when subject to a restricted feeding protocol. While mice lacking MC3R fed ad libitum exhibit normal to moderate hyperinsulinemia, when subjected to a restricted protocol they develop hyperglycemia, glucose intolerance, and dyslipidemia. Collectively, our data suggest that the central nervous melanocortin system is a point convergence in the control of energy balance and the expression of rhythms anticipating nutrient intake.
Collapse
Affiliation(s)
- Karima Begriche
- Department of Metabolism and Aging, The Scripps Research Institute-Florida, Jupiter, FL 33458, USA
| | | | | |
Collapse
|
178
|
Velásquez DA, Martínez G, Romero A, Vázquez MJ, Boit KD, Dopeso-Reyes IG, López M, Vidal A, Nogueiras R, Diéguez C. The central Sirtuin 1/p53 pathway is essential for the orexigenic action of ghrelin. Diabetes 2011; 60:1177-85. [PMID: 21386086 PMCID: PMC3064091 DOI: 10.2337/db10-0802] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Ghrelin is a stomach-derived peptide that increases food intake through the activation of hypothalamic AMP-activated protein kinase (AMPK). However, the molecular mechanisms initiated by the activation of the ghrelin receptor, which in turn lead to AMPK activation, remain unclear. Sirtuin 1 (SIRT1) is a deacetylase activated in response to calorie restriction that acts through the tumor suppressor gene p53. We tested the hypothesis that the central SIRT1/p53 pathway might be mediating the orexigenic action of ghrelin. RESEARCH DESIGN AND METHODS SIRT1 inhibitors, such as Ex527 and sirtinol, and AMPK activators, such as AICAR, were administered alongside ghrelin in the brain of rats and mice (wild-type versus p53 knockout [KO]). Their hypothalamic effects on lipid metabolism and changes in transcription factors and neuropeptides were assessed by Western blot and in situ hybridization. RESULTS The central pretreatment with Ex527, a potent SIRT1 inhibitor, blunted the ghrelin-induced food intake in rats. Mice lacking p53, a target of SIRT1 action, failed to respond to ghrelin in feeding behavior. Ghrelin failed to phosphorylate hypothalamic AMPK when rats were pretreated with Ex527, as it did in p53 KO mice. It is noteworthy that the hypothalamic SIRT1/p53 pathway seems to be specific for mediating the orexigenic action of ghrelin, because central administration of AICAR, a potent AMPK activator, increased food intake in p53 KO mice. Finally, blockade of the central SIRT1 pathway did not modify ghrelin-induced growth hormone secretion. CONCLUSIONS Ghrelin specifically triggers a central SIRT1/p53 pathway that is essential for its orexigenic action, but not for the release of growth hormone.
Collapse
Affiliation(s)
- Douglas A. Velásquez
- Department of Physiology, School of Medicine—Instituto de Investigaciones Sanitarias (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Gloria Martínez
- Department of Physiology, School of Medicine—Instituto de Investigaciones Sanitarias (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Amparo Romero
- Department of Physiology, School of Medicine—Instituto de Investigaciones Sanitarias (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María J. Vázquez
- Department of Physiology, School of Medicine—Instituto de Investigaciones Sanitarias (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Katia D. Boit
- Department of Physiology, School of Medicine—Instituto de Investigaciones Sanitarias (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), University of Santiago de Compostela, Santiago de Compostela, Spain
- Catarinense Institut of Environmental Research and Human Development, Capivari de Baixo, Santa Catarina, Brazil
| | - Iria G. Dopeso-Reyes
- Department of Cell Biology and Ecology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel López
- Department of Physiology, School of Medicine—Instituto de Investigaciones Sanitarias (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Anxo Vidal
- Department of Physiology, School of Medicine—Instituto de Investigaciones Sanitarias (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ruben Nogueiras
- Department of Physiology, School of Medicine—Instituto de Investigaciones Sanitarias (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), University of Santiago de Compostela, Santiago de Compostela, Spain
- Corresponding authors: Ruben Nogueiras, , and Carlos Diéguez,
| | - Carlos Diéguez
- Department of Physiology, School of Medicine—Instituto de Investigaciones Sanitarias (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), University of Santiago de Compostela, Santiago de Compostela, Spain
- Corresponding authors: Ruben Nogueiras, , and Carlos Diéguez,
| |
Collapse
|
179
|
Abstract
The function of adult tissue-specific stem cells declines with age, which may contribute to the physiological decline in tissue homeostasis and the increased risk of neoplasm during aging. Old stem cells can be 'rejuvenated' by environmental stimuli in some cases, raising the possibility that a subset of age-dependent stem cell changes is regulated by reversible mechanisms. Epigenetic regulators are good candidates for such mechanisms, as they provide a versatile checkpoint to mediate plastic changes in gene expression and have recently been found to control organismal longevity. Here, we review the importance of chromatin regulation in adult stem cell compartments. We particularly focus on the roles of chromatin-modifying complexes and transcription factors that directly impact chromatin in aging stem cells. Understanding the regulation of chromatin states in adult stem cells is likely to have important implications for identifying avenues to maintain the homeostatic balance between sustained function and neoplastic transformation of aging stem cells.
Collapse
Affiliation(s)
- E A Pollina
- Department of Genetics, Stanford University, CA, USA
| | | |
Collapse
|
180
|
Ramadori G, Coppari R. Does hypothalamic SIRT1 regulate aging? Aging (Albany NY) 2011; 3:325-328. [PMID: 21464518 PMCID: PMC3091526 DOI: 10.18632/aging.100311] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 03/18/2011] [Indexed: 05/30/2023]
Abstract
In virtually all organisms, life expectancy is profoundly affected by caloric intake. For example, dietary restriction (DR; a feeding regimen of fewer calories compared to the ad libitum level without causing malnutrition) has been shown to lengthen, whereas hypercaloric (HC) diet feeding to shorten, lifespan. Recent findings in invertebrates indicate that specialized groups of cells (e.g.: metabolic-sensing neurons) detect changes in caloric intake and convey energy-status-variation signals to other cells in the body to regulate lifespan. In mammals, whether metabolic-sensing neurons govern aging in a cell-non-autonomous fashion is unknown. Yet, this is a captivating and testable hypothesis.
Collapse
Affiliation(s)
- Giorgio Ramadori
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Roberto Coppari
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Faculty of Medicine, Universita’ Politecnica delle Marche, Ancona 60020, Italy
| |
Collapse
|
181
|
Maiese K, Chong ZZ, Shang YC, Wang S. Translating cell survival and cell longevity into treatment strategies with SIRT1. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2011; 52:1173-85. [PMID: 22203920 PMCID: PMC3253557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The sirtuin SIRT1, a class III NAD(+)-dependent protein histone deacetylase, is present throughout the body that involves cells of the central nervous system, immune system, cardiovascular system, and the musculoskeletal system. SIRT1 has broad biological effects that affect cellular metabolism as well as cellular survival and longevity that can impact both acute and chronic disease processes that involve neurodegenerative disease, diabetes mellitus, cardiovascular disease, and cancer. Given the intricate relationship SIRT1 holds with a host of signal transduction pathways ranging from transcription factors, such as forkhead, to cytokines and growth factors, such as erythropoietin, it becomes critical to elucidate the cellular pathways of SIRT1 to safely and effectively develop and translate novel avenues of treatment for multiple disease entities.
Collapse
Affiliation(s)
- K Maiese
- Department of Neurology and Neurosciences, Cancer Center, F 1220, UMDNJ - New Jersey Medical School, Newark, NJ, USA.
| | | | | | | |
Collapse
|
182
|
Abstract
Changes in physical activities and feeding habits have transformed the historically rare disease of obesity into a modern metabolic pandemic. Obesity occurs when energy intake exceeds energy expenditure over time. This energy imbalance significantly increases the risk for cardiovascular disease and type 2 diabetes mellitus and as such represents an enormous socioeconomic burden and health threat. To combat obesity, a better understanding of the molecular mechanisms and neurocircuitries underlying normal body weight homeostasis is required. In the 1940s, pioneering lesion experiments unveiled the importance of medial and lateral hypothalamic structures. In the 1980s and 1990s, several neuropeptides and peripheral hormones critical for appropriate feeding behavior, energy expenditure, and hence body weight homeostasis were identified. In the 2000s, results from metabolic analyses of genetically engineered mice bearing mutations only in selected neuronal groups greatly advanced our knowledge of the peripheral/brain feedback-loop modalities by which central neurons control energy balance. In this review, we will summarize these recent progresses with particular emphasis on the biochemical identities of hypothalamic neurons and molecular components underlying normal appetite, energy expenditure, and body weight homeostasis. We will also parse which of those neurons and molecules are critical components of homeostatic adaptive pathways against obesity induced by hypercaloric feeding.
Collapse
Affiliation(s)
- Claudia R Vianna
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Room Y6-220C, Dallas, Texas 75390-9077, USA
| | | |
Collapse
|
183
|
Toiber D, Sebastian C, Mostoslavsky R. Characterization of nuclear sirtuins: molecular mechanisms and physiological relevance. Handb Exp Pharmacol 2011; 206:189-224. [PMID: 21879451 DOI: 10.1007/978-3-642-21631-2_9] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sirtuins are protein deacetylases/mono-ADP-ribosyltransferases found in organisms ranging from bacteria to humans. This group of enzymes relies on nicotinamide adenine dinucleotide (NAD(+)) as a cofactor linking their activity to the cellular metabolic status. Originally found in yeast, Sir2 was discovered as a silencing factor and has been shown to mediate the effects of calorie restriction on lifespan extension. In mammals seven homologs (SIRT1-7) exist which evolved to have specific biological outcomes depending on the particular cellular context, their interacting proteins, and the genomic loci to where they are actively targeted. Sirtuins biological roles are highlighted in the early lethal phenotypes observed in the deficient murine models. In this chapter, we summarize current concepts on non-metabolic functions for sirtuins, depicting this broad family from yeast to mammals.
Collapse
Affiliation(s)
- Debra Toiber
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
184
|
Satoh A, Stein L, Imai S. The role of mammalian sirtuins in the regulation of metabolism, aging, and longevity. Handb Exp Pharmacol 2011; 206:125-62. [PMID: 21879449 PMCID: PMC3745303 DOI: 10.1007/978-3-642-21631-2_7] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Ever since the discovery of sirtuins a decade ago, interest in this family of NAD-dependent deacetylases has exploded, generating multiple lines of evidence implicating sirtuins as evolutionarily conserved regulators of lifespan. In mammals, it has been established that sirtuins regulate physiological responses to metabolism and stress, two key factors that affect the process of aging. Further investigation into the intimate connection among sirtuins, metabolism, and aging has implicated the activation of SIRT1 as both preventative and therapeutic measures against multiple age-associated disorders including type 2 diabetes and Alzheimer's disease. SIRT1 activation has clear potential to not only prevent age-associated diseases but also to extend healthspan and perhaps lifespan. Sirtuin activating compounds and NAD intermediates are two promising ways to achieve these elusive goals.
Collapse
Affiliation(s)
| | | | - Shin Imai
- Department of Developmental Biology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
185
|
Kumar KK, Tung S, Iqbal J. Bone loss in anorexia nervosa: leptin, serotonin, and the sympathetic nervous system. Ann N Y Acad Sci 2010; 1211:51-65. [PMID: 21062295 DOI: 10.1111/j.1749-6632.2010.05810.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Anorexia nervosa (AN), a disorder characterized by the refusal to sustain a healthy weight, has the highest mortality of any psychiatric disorder. This review presents a model of AN that ties together advances in our understanding of how leptin, serotonin, and hypogonadism are brought about in AN and how they influence bone mass. Serotonin (5-hydroxytryptamine) is a key regulator of satiety and mood. The primary disturbance in AN results from alterations in serotonin signaling. AN patients suffer from serotonergic hyperactivity of Htr1a-dependent pathways that causes dysphoric mood and promotes restrictive behavior. By limiting carbohydrate ingestion, anorexics decrease their serotonin levels. Reduced serotonergic signaling in turn suppresses appetite through Htr1a/2b, decreases dysphoric mood through Htr1a/2a, and activates the sympathetic nervous system (SNS) through Htr2c receptors in the ventromedial hypothalamus. Activation of the SNS decreases bone mass through β2-adrenergic signaling in osteoblasts. Additional topics reviewed here include osteoblastic feedback of metabolism in anorexia, mechanisms whereby dietary changes exacerbate bone loss, the role of caloric restriction and Sirt1 in bone metabolism, hypothalamic hypogonadism's effects on bone mass, and potential treatments.
Collapse
Affiliation(s)
- Kevin K Kumar
- Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | |
Collapse
|
186
|
Abstract
The family of protein deacetylases represented by yeast Sir2 has been the focus of intense investigation because of the longevity activity of Sir2 in yeast, worms and flies. Research in mammals has mainly focused on SIRT1, the closest homologue of Sir2. Emerging evidence from mouse models is yielding a sharper picture, in which SIRT1 is a potent protector from ageing-associated pathologies, such as diabetes, liver steatosis, cardiovascular disease, neurodegeneration and, importantly, various types of cancer.
Collapse
Affiliation(s)
- Daniel Herranz
- Tumor Suppression Group Spanish National Cancer Research Center (CNIO) Madrid, Spain
| | - Manuel Serrano
- Tumor Suppression Group Spanish National Cancer Research Center (CNIO) Madrid, Spain
- Correspondence: Manuel Serrano, Spanish National Cancer Research Center (CNIO), 3 Melchor Fernandez Almagro street, Madrid E-28029, Spain, Tel.: +34.91.732.8000, Fax: +34.91.732.8033,
| |
Collapse
|
187
|
Abstract
The hypothalamus is the center of controlling food intake and energy expenditure by integrating information on energy status, i.e. adiposity and nutrient signals. Especially, two types of neurons in the arcuate nucleus of the hypothalamus, anorexigenic proopiomelanocortin (POMC) neurons and orexigenic agouti-related peptide (AgRP) neurons, play vital roles in regulating feeding and energy expenditure. On the other hand, insulin and leptin are hormones that control food intake via regulating POMC and AgRP expression. FoxO1 is a downstream effecter of insulin signaling and Sirt1 is an NAD(+)-dependent deacetylase, both of which have been reported to play important roles in the regulation of metabolism in various organs including liver, pancreas, muscle, adipose tissue and hypothalamus. Histological analyses revealed that FoxO1 and Sirt1 are expressed in both AgRP and POMC neurons where FoxO1 localizes to the nucleus in the fasted, while to the cytoplasm in the refed condition. In contrast, hypothalamic Sirt1 protein is decreased in the fasted condition due to increased ubiquitination of Sirt1. In rodents, overexpression of FoxO1 in the hypothalamus by adenovirus microinjection induces hyperphagia and body weight gain, and simultaneous overexpression of Sirt1 suppresses these phenotypes. FoxO1 and the transcription factor Stat3 exert opposing actions on the expression of AgRP and POMC through transcriptional squelching, and Sirt1 suppresses AgRP expression. In conclusion, we propose that FoxO1 and Sirt1 in hypothalamus are key regulators of energy homeostasis and are molecular targets for the development of new strategy of treating obesity.
Collapse
Affiliation(s)
- Tsutomu Sasaki
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, Japan
| | | |
Collapse
|